ArticlePDF Available

Two Novel Mutations in the Thyrotropin (TSH) Receptor Gene in a Child with Resistance to TSH 1

Authors:

Abstract and Figures

The TSH receptor is a G protein-coupled receptor that mediates the effects of TSH in thyroid development, growth, and synthetic function. We report here that a child with features of TSH resistance, including markedly increased serum TSH concentrations and low normal thyroid hormone levels, is a compound heterozygote for two novel mutations in the TSH receptor gene. One allele has a G to A transition corresponding to an arginine to glutamine change at codon 109 (R109Q) in the extracellular domain of the receptor. The other allele has a G to A transition corresponding to a premature termination codon at tryptophan 546 (W546X) in the fourth transmembrane segment. Each parent is heterozygous for one mutation, and both parents have normal thyroid function. Cells transiently transfected with the R109Q mutant exhibited reduced membrane binding of [125I]TSH and impaired signal transduction in response to TSH. In contrast, the W546X mutant was nonfunctional, with negligible membrane radioligand binding. Our findings indicate that a single normal TSH receptor allele is sufficient for normal thyroid function, but that the compound abnormality in the proband leads to TSH resistance.
Content may be subject to copyright.
Two Novel Mutations in the Thyrotropin (TSH) Receptor
Gene in a Child with Resistance to TSH*
R. J. CLIFTON-BLIGH†, J. W. GREGORY, M. LUDGATE, R. JOHN, L. PERSANI,
C. ASTERIA, P. BECK-PECCOZ, AND V. K. K. CHATTERJEE
Department of Medicine, University of Cambridge, Addenbrooke’s Hospital (R.J.C.-B., V.K.K.C.),
Cambridge, United Kingdom CB2 2QQ; the Departments of Child Health (J.W.G.), Pathology (M.L.),
and Medical Biochemistry (R.J.), University Hospital of Wales, Cardiff, United Kingdom CF4 4XW;
and the Institute of Endocrine Sciences, University of Milan, Ospedale Maggiore, IRCCS and Centro
Auxologico Italiano IRCCS (L.P., C.A., P.B.-P.), Milan, Italy
ABSTRACT
The TSH receptor is a G protein-coupled receptor that mediates the
effects of TSH in thyroid development, growth, and synthetic func-
tion. We report here that a child with features of TSH resistance,
including markedly increased serum TSH concentrations and low
normal thyroid hormone levels, is a compound heterozygote for two
novel mutations in the TSH receptor gene. One allele hasaGtoA
transition corresponding to an arginine to glutamine change at codon
109 (R109Q) in the extracellular domain of the receptor. The other
allele hasaGtoAtransition corresponding to a premature termi-
nation codon at tryptophan 546 (W546X) in the fourth transmem-
brane segment. Each parent is heterozygous for one mutation, and
both parents have normal thyroid function. Cells transiently trans-
fected with the R109Q mutant exhibited reduced membrane binding
of [
125
I]TSH and impaired signal transduction in response to TSH. In
contrast, the W546X mutant was nonfunctional, with negligible mem-
brane radioligand binding. Our findings indicate that a single normal
TSH receptor allele is sufficient for normal thyroid function, but that
the compound abnormality in the proband leads to TSH resistance.
(J Clin Endocrinol Metab 82: 1094–1100, 1997)
P
ITUITARY TSH is critical for thyroid development and
function, and its actions are mediated by a transmem-
brane receptor, which, together with LH/CG and FSH re-
ceptors, belongs to a subfamily of G protein-coupled recep-
tors. The principal biological effects of TSH on the thyrocyte
occur by receptor-mediated activation of G
s
a
and subse-
quent generation of intracellular cAMP (1, 2). The TSH re-
ceptor gene is located on chromosome 14 (3, 4), and the
extracellular domain is encoded by nine exons, with exon 10
coding transmembrane and intracellular portions of the re-
ceptor. In keeping with many other G protein-coupled re-
ceptors (5, 6), both gain and loss of function mutations have
been described in the TSH receptor. Gain of function muta-
tions are autosomal dominant and, when somatic, cause thy-
roid hyperfunctioning adenomas (7–13), whereas germline
inheritance leads to nonautoimmune congenital hyperthy-
roidism (14–18). Autosomal recessive inheritance of TSH
resistance caused by mutations in the TSH receptor was first
reported in three siblings who were compound heterozy-
gotes for mutations of two extracellular domain residues
(19). All three patients were euthyroid with normal serum
thyroid hormone concentrations but increased serum con-
centrations of TSH, indicating that the resistance in these
cases was partial. Each parent was heterozygous for one of
these mutations.
We describe a child with greatly increased serum TSH
concentrations and low normal thyroid hormone concentra-
tions who is a compound heterozygote for two novel mu-
tations in the TSH receptor gene, one inherited from each
parent. The mutation inherited from the mother corresponds
to a TSH receptor truncated in the fourth transmembrane
domain that is functionally inactive in vitro. The paternal
mutation lies in the extracellular segment of the receptor and
has a reduced binding affinity for TSH, resulting in impaired
signal transduction.
Case Report
The patient wasidentified after a positive resulton neonatal screening
for hypothyroidism. He is the only child of unrelated parents. He was
delivered by caesarean section at 38 weeks gestation, weighed 2.45 kg,
and had transienthypoglycemia. Thyroid function tests at 8 weeks of age
showed a serum free T
4
concentration of 10 pmol/L (normal range,
12–28 pmol/L) with a TSH of 92 mU/L (normal range, 0.44.0 mU/L).
There were no clinical features of hypothyroidism. He was commenced
on T
4
. The progress of his thyroid function tests in response to T
4
administration is shown in Fig. 1. At 12 months of age, a T
4
dose of 60
m
g suppressed serum TSH into the normal range but with raised serum
free T
4
concentrations of 40 pmol/L. While on treatment, his mother
described him as irritable; when treatment was discontinued at 2 yr of
age, his behavior became more normal. After birth, he demonstrated
catch-up growth to the 50th percentile, and thereafter his growth, bone
maturation, and development have continued to be normal without
treatment. His hearing is normal, as measured by otoacoustic emissions.
A thyroid isotope scan at 2 yr of age showed a gland of normal size and
location, with uniform tracer uptake. Thyroid ultrasonography was
normal. A perchlorate discharge test was slightly elevated at 15%. Both
patient and mother were negative for thyroid autoantibodies. Serum
electrolyte, GH, PRL, LH, FSH, PTH, and calcium concentrations were
all normal in the patient. He remains clinically euthyroid without ther-
Received November 7, 1996. Revision received December 9, 1996.
Accepted December 30, 1996.
Address all correspondence and requests for reprints to: Dr. V. K. K.
Chatterjee, Department of Medicine, University of Cambridge, Level 5,
Addenbrooke’s Hospital, Hills Road, Cambridge, United Kingdom CB2
2QQ.
* This work was supported by the Wellcome Trust (to V.K.K.C.), the
Medical Research Council (to M.L.), and Murst and CNR, Rome (to
P.B.P.).
† Commonwealth Foundation Research Scholar.
0021-972X/97/$03.00/0 Vol. 82, No. 4
Journal of Clinical Endocrinology and Metabolism Printed in U.S.A.
Copyright © 1997 by The Endocrine Society
1094
apy. His most recent thyroid function tests at 3 yr of age show a TSH
of 134 mU/L and free T
4
of 12 pmol/L. The results of a T
3
suppression
test are shown in Table 2. At this time the serum glycoprotein hormone
a
-subunit level was 2.5 ng/mL (normal range, 0.24–1.05 ng/mL), but the
molar ratio of
a
-subunit to TSH was normal at 0.17 (normal range, ,5.5).
TRH tests in both parents were normal (data not shown).
Materials and Methods
Genetic analyses
Patient lymphocytes were Epstein-Barr virus-transformed (ECACC,
Porton Down, Salisbury) and grown in RPMI 1640 supplemented with
10% FCS. Ribonucleic acid and genomic DNA were extracted from the
lymphocytes using TRIzol according to the manufacturer’s instructions
(Life Technologies, Paisley, Scotland). First strand complementary DNA
(cDNA) was synthesized using Superscript II reverse transcriptase (Life
Technologies), and nested reverse transcription-PCR was subsequently
performed with primers spanning the extracellular and transmembrane
coding regions of the TSH receptor. Genomic DNA was isolated from
peripheral blood leukocytes from each parent using standard tech-
niques. Exons of the TSH receptor were amplified from patient and
parents by PCR using recently published intronic primer sequences (20).
The forward primer in each case was tagged with the universal M13
primer sequence. Direct sequencing of the PCR products was under-
taken by cycle sequencing using dye-labeled universal 21M13 primers
(Applied Biosystems/Perkin-Elmer, Cheshire, UK) and analyzed by
electrophoresis on an ABI 373 sequencer (Applied Biosystems, Foster
City, CA). Primer sequences and PCR conditions are available on
request.
Functional studies
TSH bioactivity was measured in Chinese hamster ovary cells ex-
pressing recombinant human TSH receptor as previously described (21).
DNA fragments bearing each mutation were replaced in full-length
wild-type TSH receptor cDNA cloned into the eukaryotic expression
vector pSVL (22), and constructs were verified by sequencing. JEG3
(human choriocarcinoma) cells were grown in Optimem (Life Technol-
ogies) supplemented with 2% (vol/vol) FCS and 1% (vol/vol) penicillin,
streptomycin, fungizone (Life Technologies). Eighteen hours before
transfection, the medium was replaced with Optimem containing 2%
resin-stripped FCS. Cells were transfected by a 5-h exposure to calcium
phosphate containing
a
LUC reporter, TSH receptor expression vector,
and internal control plasmid BOS-
b
-galactosidase. Twenty-four hours
after transfection, the medium was replaced to include bovine TSH
(Sigma, Dorset, UK) or recombinant human TSH (Genzyme, West
Malling, UK) as appropriate. Sixteen hours later, cells were lysed and
assayed for luciferase and galactosidase activities. Data are the mean 6
se of at least three separate experiments performed in triplicate.
Radiolabeled ligand binding studies were performed using mem-
branes extracted from COS cells transiently transfected with the receptor
expression vectors described above. Membranes were prepared as pre-
viously described (23), and protein was quantified by the Bradford
assay. Equal amounts (30–70
m
g) of protein were incubated for 2 h with
0.5 kilobecquerels [
125
I]TSH (RSR, Cardiff, UK) in NaCl-free assay buffer
with isotonicity maintained with 280 mmol/L sucrose (23) and in the
presence of increasing amounts of unlabeled bovine TSH (Sigma, Dorset,
UK). Radiolabeled TSH bound after washing was determined by
g
-counting.
Results
The results of thyroid function tests of all family members
are shown in Table 1. The biological activity of TSH in serum
from the proband was measured in Chinese hamster ovary
cells expressing recombinant human TSH receptor, and the
ratio of bioreactivity to immunoreactivity was normal at 0.69
(normal range, 0.6–2.1). This is in contrast to a recent report
of congenital hypothyroidism caused by biologically inactive
TSH due to a mutation in the TSH
b
-subunit gene (24). Thus,
the patient demonstrated features of TSH resistance, namely
reduced circulating thyroid hormone levels together with
elevated bioactive TSH. As the endocrine abnormality ap-
peared to be confined to the thyroid, analysis of the TSH
receptor gene was undertaken. Direct sequencing of the ex-
tracellular and transmembrane-coding regions of the TSH
receptor showed that the patient was heterozygous for a
CGG to CAG mutation in exon 4 corresponding to an argi-
nine to glutamine change at codon 109 (R109Q) and was
heterozygous for a second TGG to TAG mutation in exon 10
corresponding to a premature termination at codon 546
(W546X; data not shown). The mutations were verified by
repeated sequencing of both genomic DNA and receptor
cDNA isolated from the patients’ Epstein-Barr virus-trans-
formed lymphocytes. Exons 4 and 10 were amplified and
sequenced from genomic DNA isolated from each parent,
showing that mother was heterozygous for the W546X mu-
tation, and the father was heterozygous for the R109Q
mutation.
The functional consequences of each mutation were in-
vestigated using a method similar to that employed by
Sunthornthepvarakul and colleagues (19). Expression vec-
FIG. 1. Free T
4
(F) and TSH (Ç) concentrations of the patient in
relation to T
4
dose. Normal ranges for free T
4
(dotted area) and TSH
(cross-hatched area) are shown. T
4
therapy was discontinued at 24
months of age.
TABLE 1. Thyroid function test results at diagnosis
TSH
(mU/L)
Free T
4
(pmol/L)
Free T
3
(pmol/L)
TSH receptor
mutation
Patient
a
92 10 R109Q, W546X
Mother 1.5 16.3 8.1 W546X
Father 3.9 11.2 6.2 R109Q
Normal range 0.44.0 9–20 3–7.5
a
Normal range for a neonate: TSH, 0.44.0 mU/L; free T
4
, 12–28
pmol/L.
NOVEL MUTATIONS IN THE TSH RECEPTOR GENE 1095
tors encoding wild-type or mutant receptor were transiently
cotransfected with a reporter gene consisting of the glyco-
protein hormone
a
-subunit promoter and luciferase gene
(
a
LUC) into JEG3 cells. The
a
LUC reporter is highly respon-
sive in this system due to tandemly repeated cAMP response
elements between 2146 and 2111 bp of the
a
-subunit pro-
moter (25). Each mutant demonstrated impaired signal trans-
duction in response to TSH compared to that of the wild type
(Fig. 2a). The W546X mutant did not stimulate reporter gene
activity above basal levels, whereas the R109Q mutant
showed a right-shifted dose-response profile, such that max-
imal wild-type activity was achieved at higher concentra-
tions of TSH. Similar results, indicating impaired signal
transduction, were obtained using recombinant human TSH
(data not shown). To recapitulate the parental genotypes in
vitro, either mutant was cotransfected in equal amounts with
wild-type receptor. The resulting dose-response profiles did
not differ from that seen with wild-type receptor alone,
whereas cotransfection of both mutants reproduced the ac-
tivity of transfection of R109Q alone (Fig. 2b). Thus, neither
mutant was able to dominantly inhibit the activity of wild-
type receptor, nor was any positive cooperativity between
the mutants observed. Although constitutive activation of
the cAMP cascade by unliganded TSH receptor has been
described in COS cells (7, 9, 14), such basal activity of either
wild-type or mutant TSH receptors could not be detected in
this assay. This may be due to a limitation of the luciferase
reporter system, as previous characterization of mutant TSH
receptors using this assay in COS cells also failed to show
constitutive receptor activity (19).
Binding studies with radiolabeled ligand were undertaken
using the same receptor expression vectors transiently trans-
fected in COS-7 cells (Fig. 3). The R109Q mutant showed a
binding capacity of 1140 cpm, which is 60% that of wild-type
receptor (1900 cpm), and raised EC
50
(20 vs. 7mUofthe
bovine TSH preparation we used; data not shown), consis-
tent with reduced binding affinity for TSH. When experi-
ments were performed on whole cells, the binding to R109Q,
although higher than that on W546X or vector alone, was too
low to obtain a curve, indicating very poor surface expres-
sion of the mutant (data not shown). The W546X mutant
receptor showed negligible specific binding for TSH on ei-
ther whole cells or membranes, which is probably indicative
of very little surface expression of this receptor, although the
possibility that the truncated receptor still inserts into the
membrane but fails to bind TSH through lacking C-terminal
residues cannot be excluded. Nevertheless, poor cellular ex-
pression of prematurely truncated receptors is well de-
scribed (26–28).
Persistent elevation of serum TSH concentrations in this
child (Fig. 1) has prompted concern as to whether there may
be a risk of developing pituitary autonomy. Magnetic reso-
nance imaging (MRI) performed at 2 yr of age showed two
small areas of hypoattenuation after the administration of
gadolinium (Fig. 4). A T
3
suppression test was subsequently
performed, which showed brisk reduction of TSH into the
FIG. 2. Function of wild-type and mutant TSH receptors. a, Activation of
a
LUC by wild-type or mutant receptors transfected individually. Cells
were transfected with 500 ng reporter, 100 ng receptor expression vector, and 100 ng BOS-
b
-galactosidase. Luciferase activity was determined
after incubation with 0–100 mU/mL bovine TSH and normalized for transfection efficiency using
b
-galactosidase activity. The mean (6SE)
responses to bovine TSH are expressed relative to the maximum response obtained with wild-type receptor, which was approximately 10-fold.
b, Coexpression of wild type with each mutant receptor expression vector. Cells were transfected with reporter and reference plasmids, as
described in a, and 50 ng receptor expression vectors in the indicated combinations. For comparison 50 ng R109Q vector were transfected
individually with 50 ng pSVL to correct for DNA concentration. Hormone-dependent activation after incubation with 0–100 mU/mL bovine TSH
was determined as described above. Where values are less than 10% of the mean, error bars have been omitted for clarity. The combinations
of wild-type and each mutant receptor reflect the heterozygous nature of each parent (mother and father), whereas the combination of both
mutant receptors represents the compound state of the proband.
1096 CLIFTON-BLIGH ET AL.
JCE&M1997
Vol 82 No 4
normal range after the administration of T
3
(Table 2). Cre-
atine kinase, cholesterol, and triglyceride levels showed a
normal response to exogenous T
3
, although there was little
change in serum sex hormone-binding globulin or alkaline
phosphatase levels. Overall, these results suggest normal
pituitary and peripheral tissue responsiveness to T
3
in the
patient. Sequencing of the thyroid hormone receptor
b
gene
in the proband was also normal (data not shown).
Discussion
We have described a child with persistent hyperthyro-
tropinemia together with normal serum thyroid hormone
concentrations associated with two novel mutations in the
TSH receptor gene. When tested individually, each mutant
receptor showed impaired function in transfection studies.
Although the R109Q mutant receptor showed only mild im-
pairment, clinical data from the proband suggest that this
mutation results in markedly abnormal thyroid function. It
is possible that the limited dynamic range (10-fold) of the
transfection assay may underestimate the in vivo conse-
quences of the R109Q mutation. Neither mutant receptor
affected wild-type receptor function when coexpressed,
which is concordant with the observation that both parents
have normal thyroid function. Coexpression of both mutant
receptors results in some residual functional activity, which
correlates with the partial TSH resistance seen in the patient.
As expected from the autosomal recessive nature of this
disorder, neither mutant was able to dominantly inhibit the
function of wild-type receptor.
Our case is the second recorded example of loss of function
mutations in the TSH receptor gene (19). All cases described
hitherto have been euthyroid, indicating that TSH resistance
in each case is only partial, such that the elevated TSH levels
are able to stimulate adequate thyroid hormone secretion. In
contrast, the phenotype of more severe TSH resistance is
represented by the recessively inherited hyt/hyt hypothyroid
mouse (29). Here, fetal onset of profound hypothyroidism is
associated with greatly elevated TSH levels. The homozy-
gous mutant thyroid gland is hypoplastic and demonstrates
diminished follicular size and reduced colloid. Recently, a
point mutation in the TSH receptor gene was identified in
this mouse, corresponding to a Pro to Leu change at codon
FIG. 4. Pituitary MRI scan in the proband at 2 yr of age. Two areas of hypoattenuation after the administration of gadolinium are indicated
(arrows). The contours of the gland are normal.
TABLE 2. Thyroid function and indexes of hormone action during
aT
3
suppression test
Results Basal
T
3
,15
m
g for
3 days (% of basal)
T
3
,30
m
g for
3 days (% of basal)
Free T
4
(pmol/L) 12.0 11.2 (93) 9.8 (82)
Free T
3
(pmol/L) 6.4 9.9 (155) 10.3 (161)
TSH (mU/L) 134.8 56.4 (42) 9.5 (7)
Sex hormone-binding
globulin (nmol/L)
114 110 (96) 103 (90)
Alkaline phosphatase
(IU/L)
212 209 (99) 184 (87)
Cholesterol (mmol/L) 5.8 4.8 (83) 4.0 (69)
Creatine kinase (IU/L) 186 135 (73) 132 (71)
Triglyceride (mmol/L) 1.3 0.6 (46) 0.7 (54)
FIG. 3. A representative experiment showing binding of [
125
I]TSH to
membrane-associated TSH receptors. COS-7 cells were transfected
with the same receptor expression plasmids as in Fig. 2, and mem-
branes were incubated with [
125
I]TSH and 0–100 mU/mL unlabeled
bovine TSH. Mean (6SE) binding values of triplicate determinations,
expressed as counts per min, are shown.
NOVEL MUTATIONS IN THE TSH RECEPTOR GENE 1097
556 in the transmembrane region, which abolished TSH bind-
ing and response to TSH in vitro (30).
The locations of loss of function mutations in the TSH
receptor identified to date are shown in Fig. 5. Study of these
mutations has defined some important residues for TSH
binding and receptor function. The previous report indicated
that mutation of proline at codon 162 to alanine retained
some biological activity, whereas mutation of isoleucine at
codon 167 to asparagine virtually abolished signal transduc-
tion (19). A structural model for the hormone-binding site
has been proposed on the basis of similarity between the
extracellular domain of the TSH receptor and the crystal
structure of the ribonuclease inhibitor (31, 32). From this
model, the arginine at codon 109, which is mutated in our
case, may be expected to project into solution, contributing
to the TSH binding cavity. This hypothesis is confirmed by
studies indicating that mutation of this residue to glutamine
interferes with TSH binding. Both arginine at codon 109 and
tryptophan at codon 546 are conserved in the TSH receptor
from a number of species. Furthermore, tryptophan at codon
546 in the TSH receptor is conserved at homologous positions
in both the LH/CG and FSH receptors (33, 34). Curiously, in
the LH/CG receptor, the residue homologous to arginine 109
is glutamine. However, the transfected R109Q mutant TSH
receptor did not mediate signal transduction in response to
human LH (data not shown).
Our case contributes two more important findings. First,
it describes the only occurrence hitherto of a TSH receptor
with a premature termination codon within its serpentine
portion, resulting in a biologically inactive product. The
mother of the proband is thus effectively hemizygous for
functional TSH receptors, suggesting that only a single nor-
mal TSH receptor allele is required to sustain normal thyroid
function. Second, TSH levels in the proband are the highest
of those reported to date. Elevated serum TSH levels are
characteristic of TSH resistance and probably represent re-
setting of the pituitary threshold for TSH suppression by
thyroid hormones (19), although the mechanism remains
obscure. In keeping with this, there is no evidence for resis-
tance to thyroid hormones in our patient. The occurrence of
possible pituitary abnormalities on a MRI scan in our case in
the context of persistently high TSH levels was disturbing,
although the significance of these findings remains uncer-
tain. The evidence of brisk inhibition of serum TSH levels
FIG. 5. Location of loss of function mutations in the TSH receptor structure. Mutations of proline at codon 162, isoleucine at codon 167, and
proline at codon 556 have been identified previously (19, 30). The R109Q and W546X mutations are arrowed.
1098 CLIFTON-BLIGH ET AL.
JCE&M1997
Vol 82 No 4
during a T
3
suppression test mitigates against pituitary au-
tonomy. It is also encouraging that the secretion of other
anterior pituitary hormones, the
a
-subunit/TSH molar ratio,
and the growth pattern are all normal. Nevertheless, en-
largement of the sella has been shown to occur in long term
juvenile and untreated congenital hypothyroidism (35, 36),
which may be proportional to the degree of TSH elevation
(36). Moreover, thyrotrope hyperplasia is evident at autopsy
in long term primary hypothyroidism (37). It is known from
animal studies that prolonged hypothyroidism may ulti-
mately lead to thyrotrope neoplasia (38). We intend to follow
the pituitary changes closely, with further imaging at regular
intervals.
It is clear that activating mutations of the TSH receptor
increase both thyrocyte growth and function (39). In contrast,
impaired TSH receptor function has not yet been associated
with disordered thyroid development in humans. A role for
TSH in thyroid ontogeny is suggested by evidence that thy-
roid development is arrested in late gestation in mice ho-
mozygous for a knockout of the glycoprotein hormone
a
-subunit common to TSH, LH, and FSH (40), and that sim-
ilarly in humans, thyroid dysgenesis has been reported in
some cases of TSH deficiency caused by homozygous mu-
tations in TSH
b
-subunit gene (41, 42). By analogy with the
hyt/hyt hypothyroid mouse, we speculate that a combination
of severe loss of function TSH receptor mutations will be
found in some cases of thyroid dysgenesis. However, it is
also relevant to note that cases of congenital hypothyroidism
and TSH hyporesponsiveness have been described in which
the TSH receptor gene is reportedly normal (43, 44), sug-
gesting that the molecular basis of athyreosis is likely to be
heterogeneous. Partial TSH resistance with normal gland
development, low or normal thyroid hormone concentra-
tions, and elevated bioactive TSH levels may be a more
readily identifiable entity. The study of such cases will con-
tinue to provide valuable insights into the function of the
TSH receptor.
Acknowledgment
The authors are indebted to Prof. G. Vassart for providing the wild-
type TSH receptor cDNA cloned in pSVL.
Note Added In Proof
Since the submission of this manuscript, de Roux et al. have reported
compound heterozygosity for the W546X and another mutation in the
TSH receptor in a case of TSH resistance. N. de Roux, M. Misrahi, R.
Brauner, M. Houang, J. C. Carel, M. Granier, Y. Le Bouc, N. Ghinea, A.
Boumedienne, J. E. Toublanc, E. Milgrom. 1996 Four families with loss
of function mutations of the thyrotropin receptor. J Clin Endocrinol
Metab. 81:42294235.
References
1. Vassart G, Dumont JE. 1992 The thyrotropin receptor and the regulation of
thyrocyte function and growth. Endocr Rev. 13:596611.
2. Tonacchera M, Van Sande J, Parma J, et al. 1996 TSH receptor and disease.
Clin Endocrinol (Oxf). 44:621–633.
3. Libert F, Passage E, Lefort, et al. 1990 Localization of human thyrotropin
receptor gene to chromosome region 14q31 by in situ hybridization. Cytogen
Cell Genet. 54:82–83.
4. Gross B, Misrahi M, Sar S, Milgrom E. 1991 Composite structure of the human
thyrotropin receptor gene. Biochem Biophys Res Commun. 177:679687.
5. Pearce S, Trump D. 1995 G-Protein-coupled receptors in endocrine disease. Q
J Med. 88:3–8.
6. Shenker A. 1995 G protein-coupled receptor structure and function–the impact
of disease-causing mutations. Balliere Clin Endocrinol Metab. 9:427–451.
7. Parma J, Duprez L, Van Sande J, et al. 1993 Somatic mutations in the thy-
rotropin receptor gene cause hyperfunctioning thyroid adenomas. Nature.
365:649651.
8. Porcellini A, Ciullo I, Laviola L, Amabile G, Fenzi G, Avvedimento VE. 1994
Novel mutations of thyrotropin receptor gene in thyroid hyperfunctioning
adenomas. Rapid identification by fine needle aspiration biopsy. J Clin En-
docrinol Metab. 79:657–661.
9. Paschke R, Tonacchera M, Van Sande J, Parma J, Vassart G. 1994 Iden-
tification and functional characterization of two new somatic mutations
causing constitutive activation of the thyrotropin receptor in hyperfunc-
tioning autonomous adenomas of the thyroid. J Clin Endocrinol Metab.
79:1785–1789.
10. Takeshita A, Nagayama Y, Yokoyama N, et al. 1995 Rarity of oncogenic
mutations in the thyrotropin receptor of autonomously functioning thyroid
nodules in Japan. J Clin Endocrinol Metab. 80:2607–2611.
11. Parma J, Van Sande J, Swillens S, Tonacchera M, Dumont J, Vassart G. 1995
Somatic mutations causing constitutive activity of the thyrotropin receptor are
the major cause of hyperfunctioning thyroid adenomas: identification of ad-
ditional mutations activating both the cyclic adenosine 39,59-monophosphate
and inositol phosphate-Ca
21
cascades. Mol Endocrinol. 9:725–733.
12. Russo D, Arturi F, Wicker R, et al. 1995 Genetic alterations in thyroid hy-
perfunctioning adenomas. J Clin Endocrinol Metab. 80:1347–1351.
13. Russo D, Arturi F, Suarez HG, et al. 1996 Thyrotropin receptor gene alter-
ations in thyroid hyperfunctioning adenomas. J Clin Endocrinol Metab.
81:1548–1551.
14. Duprez L, Parma J, Van Sande J, et al. 1994 Germline mutations in the
thyrotropin receptor gene cause non-autoimmune autosomal dominant hy-
perthyroidism. Nat Genet. 7:396401.
15. Kopp P, Van Sande J, Parma J, et al. 1995 Brief report: congenital hyperthy-
roidism caused by a mutation in the thyrotropin receptor gene. N Engl J Med.
332:150–154.
16. Van Sande J, Parma J, Tonacchera M, Swillens S, Dumont J, Vassart G. 1995
Somatic and germline mutations of the TSH receptor gene in thyroid diseases.
J Clin Endocrinol Metab. 80:2577–2585.
17. Tonacchera M, Van Sande J, Cetani F, et al. 1996 Functional characteristics of
three new germline mutations of the thyrotropin receptor gene causing au-
tosomal dominant toxic thyroid hyperplasia. J Clin Endocrinol Metab.
18:547–554.
18. De Roux N, Polak M, Couet J, et al. 1996 A neomutation of the thyroid-
stimulating hormone receptor in a severe neonatal hyperthyroidism. J Clin
Endocrinol Metab. 81:2023–2026.
19. Sunthornthepvarakul T, Gottschalk ME, Hayashi Y, Refetoff S. 1995 Brief
report: resistance to thyrotropin caused by mutations in the thyrotropin-re-
ceptor gene. N Engl J Med. 332:155–160.
20. De Roux N, Misrahi M, Chatelain N, Gross B, Milgrom E. 1996 Microsatellites
and PCR primers for genetic studies and genomic sequencing of the human
TSH receptor gene. Mol Cell Endocrinol. 117:253–256.
21. Persani L, Tonacchera M, Beck-Peccoz P et al. 1993 Measurement of cAMP
accumulation in Chinese hamster ovary cells transfected with the recombinant
human TSH receptor (CHO-R): a new bioassay for human thyrotropin. J
Endocrinol Invest. 16:511–519.
22. Libert F, Lefort A, Gerard C, et al. 1989 Cloning, sequencing and expression
of the human thyrotropin (TSH) receptor: evidence for binding of autoanti-
bodies. Biochem Biophys Res Commun. 165:1250–1255.
23. Costagliola S, Swillens S, Niccoli P, Dumont J, Vassart G, Ludgate M. 1992
Binding assay for thyrotropin receptor autoantibodies using the recombinant
receptor protein. J Clin Endocrinol Metab. 75:1540–1544.
24. Medeiros-Neto G, Herodotou DT, Rajan S, et al. 1996 A circulating, biolog-
ically inactive thyrotropin caused by a mutation in the beta subunit gene. J Clin
Invest. 97:1250–1256.
25. Silver BJ, Bokar JA, Virgin JB, Vallen EA, Milsted A, Nilson JH. 1987 Cyclic
AMP regulation of the human glycoprotein hormone
a
-subunit gene is me-
diated by an 18-base-pair element. Proc Natl Acad Sci. USA. 84:2198–2202.
26. Brawerman G. 1989 mRNA decay: finding the right targets. Cell. 57:9–10.
27. Marcelli M, Tilley WD, Wilson CM, Griffin JE, Wilson JD, McPhaul MJ. 1990
Definition of the human androgen receptor gene structure permits the iden-
tification of mutations that cause androgen resistance: premature termination
of the receptor protein at amino acid residue 558 causes complete androgen
resistance. Mol Endocrinol. 4:1105–1116.
28. Peltz SW, He F, Welch E, Jacobsen A. 1994 Nonsense-mediated mRNA decay
in yeast. Prog Nucleic Acid Res Mol Biol. 47:271–298.
29. Beamer WG, Eicher EM, Maltais LJ, Southard JL. 1981 Inherited primary
hypothyroidism in mice. Science. 212:61–63.
30. Stein SA, Oates EL, Hall CR, et al. 1994 Identification of a point mutation in
the thyrotropin receptor of the hyt/hyt hypothyroid mouse. Mol Endocrinol.
8:129–138.
31. Kobe B, Deisenhofer J. 1993 Crystal structure of porcine ribonuclease inhib-
itor, a protein with leucine-rich repeats. Nature. 366:751–756.
32. Kajava AV, Vassart G, Wodak SJ. 1995 Modelling of the three-dimensional
structure of proteins withthe typical leucine-rich repeats. Structure. 3:867–877.
NOVEL MUTATIONS IN THE TSH RECEPTOR GENE 1099
33. Frazier AL, Robbins LS, Stork PJ, Sprengel R, Segaloff DL, Cone RD. 1990
Isolation of TSH and LH/CG receptor cDNAs from human thyroid: regulation
by tissue specific splicing. Mol Endocrinol. 4:1264–76.
34. Minegish T, Nakamura K, Takakura Y, Ibuki Y, Igarashi M. 1991 Cloning and
sequencing of human FSH receptor cDNA. Biochem Biophys Res Commun.
175:1125–1130.
35. Medeiros-Neto GA, Kourides IA, Almeida F, Gomes E, Cavaliere H, Ingbar
SH. 1981 Enlargement of the sella turcica in some patients with longstanding
untreated endemic cretinism. Serum TSH, alpha, TSH-
b
, and prolactin re-
sponses to TRH. J Endocrinol Invest. 4:303–307.
36. Yamada T, Tsukui T, Ikejiri K, Yukimura Y, Kotani M. 1976 Volume of sella
turcica in normal subjects and in patients with primary hypothyroidism and
hyperthyroidism. J Clin Endocrinol Metab. 42:817–822.
37. Scheithauer BW, Kovacs K, Randall R, Ryan N. 1985 Pituitary gland in
hypothyroidism. Histologic and immunocytologic study. Arch Pathol Lab
Med. 109:499–504.
38. Furth J, Moy P, Hershman J. 1973 Thyrotropic tumor syndrome. A multiglan-
dular disease induced by sustained deficiency of thyroid hormones. Arch
Pathol. 96:217–226.
39. Paschke R, Van Sande J, Parma J, Vassart G. 1996 The TSH receptor and
thyroid diseases. Balliere Clin Endocrinol Metab. 10:9–27.
40. Kendall SK, Samuelson LC, Saunders TL, Wood RI, Camper SA. 1995 Tar-
geted disruption of the pituitary glycoprotein hormone
a
-subunit produces
hypogonadal and hypothyroid mice. Genes Dev. 9:2007–2019.
41. Dacou-Voutetakis C, Feltquate DM, Drakopoulou M, Kourides IA,
Dracopoli NC. 1990 Familial hypothyroidism caused by a nonsense mutation
in the thyroid-stimulating hormone
b
-subunit gene. Am J Hum Genet.
46:988–993.
42. Mori R, Sawai T, Kinoshita E, et al. 1991 Rapid detection of a point
mutation in thyroid-stimulating hormone
b
-subunit gene causing congen-
ital isolated thyroid-stimulating hormone deficiency. Jpn J Hum Genet.
36:313–316.
43. Takamatsu J, Nishikawa M, Horimoto M, Oshawa N. 1993 Familial unre-
sponsiveness to thyrotropin by autosomal recessive inheritance. J Clin Endo-
crinol Metab. 77:1569–1573.
44. Takeshita A, Nagayama Y, Yamashita S, et al. 1994 Sequence analysis of the
TSH receptor gene in congenital primary hypothyroidism associated with TSH
unresponsiveness. Thyroid. 4:255–259.
1100 CLIFTON-BLIGH ET AL.
JCE&M1997
Vol 82 No 4
... Clifton-Bligh et al first reported that the TSHR p.Arg109Gln variant may lead to CH, with moderate functional deterioration. 2 We identified six previous studies that reported TSHR p.Arg109Gln (Table 1). [1][2][3][4][5][6] None of them involved follow-up of patients until adolescence. ...
... Clifton-Bligh et al first reported that the TSHR p.Arg109Gln variant may lead to CH, with moderate functional deterioration. 2 We identified six previous studies that reported TSHR p.Arg109Gln (Table 1). [1][2][3][4][5][6] None of them involved follow-up of patients until adolescence. No significant differences in sex were found in the patients (males, 3; females, 4; unknown, 1). ...
... CH-associated TSHR p.Arg109Gln may be moderate in severity, as shown in previous functional analysis. 1,2 Thus far, only one case of permanent CH-associated TSHR p.Arg109Gln and p.Arg450His compound heterozygous variants has been reported in a 12-year-old Japanese boy. 1 Our case was similar to this case because the patient had orthotopic, nongoitrous, and permanent CH; however, the I-123 uptake study results were different. The characteristic clinical feature was nonclassical TSH resistance, which is characterized by paradoxical I-123 uptake elevation. ...
Article
Full-text available
Congenital hypothyroidism (CH) is a common heterogeneous endocrine disorder. The thyroid-stimulating hormone receptor gene (TSHR) is one of the major candidate genes associated with CH. Studies have investigated the possible correlations between the specific clinical features and the presence of TSHR variants. However, only a few reports have focused on the long-term follow-up of patients with CH. Here we present a case of CH-associated TSHR p.Arg109Gln and p.Arg450His rare compound heterozygous variants, with a follow-up performed until adolescence. The patient had high serum TSH levels during newborn screening. Oral administration of levothyroxine (l-T4) was initiated at 1 month of age. The ultrasonogram revealed normal thyroid morphology and blood flow. Reduced uptake of I-123 and negative perchlorate test was observed. A small amount of l-T4 remained needed although l-T4 could be steadily reduced by puberty. The patient was diagnosed with orthotopic, nongoitrous, and permanent CH. He had no nonclassical TSH resistance. Patients with the TSHR p.Arg109Gln compound heterozygous variant exhibit permanent CH with high TSH levels and normal or slightly lower fT4 levels. In the future, genotype identification could help predict the long-term prognosis and reduce the requirement for detailed examinations. More case studies are needed to determine the relationship between genetic variants and clinical features in CH.
... Individuals with only one mutant allele are largely unaffected but may have mildly elevated TSH, indicating one normal TSHR allele is sufficient for normal thyroid function. 173,174 This disorder is now known as congenital hypothyroidism or thyroid resistance. ...
... 175 Inactivating mutations are associated with impaired receptor expression or ligand binding. [172][173][174][175] There are reports that the functional impairment of TSHR correlates with the severity of the TSHR phenotype and can be associated with profound hypoplasia of the thyroid gland. 176 TSH resistance is also associated with mutations in other genes involved in thyroid development or TSHR signaling. ...
Chapter
G-protein-coupled receptors (GPCRs) form the largest family of receptors in humans, with over 800 members. These receptors mediate the effects of many hormones and convey numerous endocrine functions including growth, appetite regulation, bone development, glucose homeostasis and reproduction. Consequently, mutations in these receptors cause a spectrum of disorders and studies of these diseases has provided new insights into GPCR functions. This has led to an enhanced appreciation of the complexity of GPCR signaling achieved, in part, by the ability of receptors to couple to multiple G-protein pathways, utilization of sophisticated spatiotemporal signaling from plasma membrane and intracellular compartments, oligomerization of receptors and activation of G-protein-independent pathways. Pharmacological strategies for targeting GPCRs have become increasingly nuanced with the development of chimeric ligands targeting multiple receptors and the emergence of compounds that enable biased signaling. Future therapeutic approaches are likely to further enhance tissue and functional specificity. This article focusses on GPCRs involved in energy metabolism and growth (melanocortin, ghrelin, somatostatin, GLP-1 and GIP receptors); calcium homeostasis and skeletal development (calcium-sensing and parathyroid hormone receptors); thyroid function (TSH receptor); and reproduction (GnRHR, LHR, FSHR, KISS1R). Mutations in these GPCRs contribute to diverse human disorders including severe obesity, hyper/hypocalcaemia, skeletal dysplasia, hyper/hypothyroidism, precocious puberty, and delayed sexual development, and many are targets for currently approved drugs. For each receptor an understanding of the biological function, signal transduction mechanisms and pathophysiological consequences of human mutations will be explained, and current and future therapeutic strategies for targeting each receptor described.
... NGS identified a total of 24 variants in 14 patients after removing unselected variants. As shown in Table 3, the variants were distributed in 15 sites (six novel sites and nine previously reported sites) of five genes [6,[20][21][22][23][24][25][26]. Eight, five, eight, two, and one variants were found to be potentially pathogenic in DUOX2, TSHR, TG, UBR1, and TPO genes, respectively. ...
Article
Congenital hypothyroidism (CH) is considered the most common congenital endocrine disorder of genetic origin. Next generation sequencing (NGS) is the standard method for identifying genetic mutations, but it is an expensive and complex technique. Therefore, we propose to use Sanger sequencing to identify selected variants of the four most common CH-causative genes: DUOX2, TG, TSHR, and PAX8. To analyze the performance of Sanger sequencing, we compared its variant detection ability with that of a CH NGS panel containing 53 genes. We performed Sanger sequencing of selected variants and panel NGS analysis of 25 Japanese patients with CH. Sanger sequencing identified nine variants in seven patients, while NGS identified 24 variants in 14 patients. Of these, eight, five, eight, two, and one were found to be potentially pathogenic in DUOX2, TSHR, TG, UBR1, and TPO genes, respectively. The percentage of detectable variants using Sanger sequencing compared with NGS was 37.5% (9/24 variants), whereas the percentage of detectable cases carrying variants using Sanger sequencing compared with NGS was 50% (7/14 patients). We proposed a system for screening commonly identified CH-related variants by Sanger sequencing. Sanger sequencing could therefore identify about a third of CH-causative variants, so is considered an effective and efficient form of pre-screening before NGS.
Article
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors, with many GPCRs having crucial roles in endocrinology and metabolism. Cryogenic electron microscopy (cryo-EM) has revolutionized the field of structural biology, particularly regarding GPCRs, over the past decade. Since the first pair of GPCR structures resolved by cryo-EM were published in 2017, the number of GPCR structures resolved by cryo-EM has surpassed the number resolved by X-ray crystallography by 30%, reaching >650, and the number has doubled every ~0.63 years for the past 6 years. At this pace, it is predicted that the structure of 90% of all human GPCRs will be completed within the next 5-7 years. This Review highlights the general structural features and principles that guide GPCR ligand recognition, receptor activation, G protein coupling, arrestin recruitment and regulation by GPCR kinases. The Review also highlights the diversity of GPCR allosteric binding sites and how allosteric ligands could dictate biased signalling that is selective for a G protein pathway or an arrestin pathway. Finally, the authors use the examples of glycoprotein hormone receptors and glucagon-like peptide 1 receptor to illustrate the effect of cryo-EM on understanding GPCR biology in endocrinology and metabolism, as well as on GPCR-related endocrine diseases and drug discovery.
Chapter
The thyroid axis is essential for the proper in and ex utero somatic and neurological development of the fetus and infant. Thyroid axis homeostasis is tightly regulated through a number of stimulatory and inhibitory mechanisms that involve the hypothalamus, the anterior pituitary gland, the thyroid gland, and peripheral tissues. Thyroid hormone, mainly T3, has both genomic and nongenomic pleiotropic effects in a variety of peripheral tissues. It increases flux through oxidative pathways, promotes growth and development, enhances the development and function of the nervous system, increases cardiac function, and permits a normal reproductive function. In utero impairment in the thyroid developmental process leads to a variety of dysfunction from thyroid dysgenesis, dyshormonogenesis, and central hypothyroidism. Molecular biology now permits novel approaches for defining the nature of the mutation involved and better management of congenital thyroid abnormalities. This chapter briefly covers the ontogeny of the thyroid axis, thyroid hormone metabolism, and the mechanisms of action before addressing the laboratory evaluation of thyroid function from the perinatal period through adolescence in health and disease.
Chapter
This chapter describes basic principles of receptor action including definitions of agonists, antagonists, inverse agonists, including basic pharmacology of receptor binding, activation and inhibition. All classes of receptors relevant to endocrinology are described, organized by receptor class, and sometimes subclass. Each class/subclass has a general description of receptor structure, expression, subcellular localization, binding, activation, downstream signaling, and deactivation. For a given class of receptors, nonendocrine receptors and ligands of the same class may be described where relevant. For example, in the description of G protein–coupled receptors the variety of ligands is described, both endocrine and nonendocrine including light, odorants and pheromones, small molecules, and proteins. Germline mutations of each receptor (whether activating or inactivating) are described along with the resulting endocrine disorder and its phenotype. In some cases germline mutations causing syndromes that do not have an endocrine phenotype are listed but not fully described. Genotype phenotype correlations are discussed where relevant. Treatment of each disorder may be briefly described but is not the focus of this chapter.
Article
Full-text available
Neonatal screening in Macedonia detects congenital hypothyroidism (CH) with an incidence of 1 in 1585 and more than 50% cases exhibit a normally-located gland-in-situ (GIS). Monogenic mutations causing dyshormonogenesis may underlie GIS CH; additionally, a small proportion of thyroid hypoplasia has a monogenic cause, such as TSHR and PAX8 defects. The genetic architecture of Macedonian CH cases has not previously been studied. We recruited screening-detected, non-syndromic GIS CH or thyroid hypoplasia cases (n=40) exhibiting a spectrum of biochemical thyroid dysfunction ranging from severe permanent to mild transient CH and including 11 familial cases. Cases were born at term, with birth weight >3000g and thyroid morphologies included goitre (n=11), thyroid hypoplasia (n=6) and apparently normal-sized thyroid. A comprehensive, phenotype-driven, Sanger sequencing approach was used to identify genetic mutations underlying CH, by sequentially screening known dyshormonogenesis-associated genes and TSHR in GIS cases and TSHR and PAX8 in cases with thyroid hypoplasia. Potentially pathogenic variants were identified in fourteen cases of which four were definitively causative; we also detected digenic variants in three cases. Seventeen variants (nine novel), were identified in TPO (n=4), TG (n=3), TSHR (n=4), DUOX2 (n=4), and PAX8 (n=2). The relatively low mutation frequency suggests that factors other than recognized monogenic causes (oligogenic variants, environmental factors or novel genes) may contribute to GIS CH in this region. Future non-hypothesis driven, next generation sequencing studies are required to confirm these findings.
Article
The thyroid stimulating hormone receptor (TSHR) mutation database, consisting of all known TSHR mutations and their clinical characterizations, was established in 1999. The database contents are updated here with the same website (tsh-receptor-mutation-database.org). The new database contains 638 cases of TSHR mutations: 448 cases of gain of function mutations (7 novel mutations and 41 new cases for previously described mutations since its last update in 2012) and 190 cases of loss of function mutations (28 novel mutations and 31 new cases for previously described mutations since its last update in 2012). This database is continuously updated and allows for rapid validation of patient TSHR mutations causing hyper- or hypothyroidism or insensitivity to TSH.
Article
Full-text available
Congenital hypothyroidism (CH) is a heterogeneous disorder with largely unknown causes, affecting 1/3000-1/4000 new-borns. Individuals with Down syndrome have a much higher incidence of CH than the normal population, probably due to the extra copy of chromosome 21. Moreover, a girl has recently been described with CH and an interstitial deletion of proximal 21q, possibly revealing a recessive disease allele on the undeleted chromosome. To establish whether chromosome 21 is also involved in the etiology of familial cases of presumably autosomal recessive CH, we investigated 22 families with recessive CH, using 10 microsatellite markers from 21q. Linkage analysis allowing for heterogeneity revealed no signs of linkage even in a small proportion of the families, and two-point analysis of the markers made it possible to exclude the long arm of chromosome 21 from containing any major disease-causing gene.
Article
Until recently, neonatal hyperthyroidism has been considered to be related to the transplacental passage of thyroid-stimulating Ig present in the serum of the mother. We report here the case of a newborn who presented with severe hyperthyroidism, diffuse goiter, and important ocular signs (eyelid retraction and possibly proptosis). However, the absence of thyroid pathology in the parents and the lack of antithyroid antibodies in the mother and in the patient led us to suspect a nonimmune aetiology. Direct genomic sequencing of the last exon of the TSH receptor in the patient revealed a T-->C transversion yielding to a Met453-->Thr heterozygous substitution in the second transmembrane domain of the receptor. The mutation was absent in both parents. Eukaryotic expression analysis in COS-7 cells yielded a mutated receptor that produced constitutive activation of adenylate cyclase without enhancement of phospholipase C activity.
Article
We have characterized a transfected Chinese hamster ovary cell line, JP09, which expresses high levels of the human TSH receptor (TSH-R). Based on a theoretical biological activity for TSH of 40 IU/mg, JP09 has approximately 90,000 receptors per cell, having a dissociation constant of 1.64 x 10(3) mU/L or 1.47 x 10(-9) mol/L. We have used JP09 to prepare solubilized TSH-Rs which have formed the basis of a binding assay for thyroid-binding inhibiting immunoglobulins in unfractionated sera. We have compared the JP09 assay with the TRAK assay (which is based on solubilized porcine TSH-R) and found a highly positive correlation between the two assays, r = 0.83 P < 0.0001, in 55 sera from patients with autoimmune thyroid disease. JP09 can be adapted to growth in suspension culture, permitting large scale production. The tracer in the assay is bovine [125I]TSH; surprisingly, despite the use of a hTSH-R, hTSH had no effect on the binding of the tracer up to 10(3) mU/L and only a minor effect at 10(4) mU/L.
Article
Under physiological circumstances, thyrotropin (TSH) is the primary hormone that controls thyroid function and growth. TSH acts by binding to its receptor at the basolateral membrane of thyroid follicular cells. The TSH receptor is a member of the large family of G protein-coupled receptors, which share a similar structural pattern: seven transmembrane segments connected by three extra and three intracellular loops. Together with the receptors for other glycoprotein hormones LH/CG and FSH, the TSH receptor has a long aminoterminal domain that has been shown to encode the specificity for hormone recognition and binding. The G protein-coupled receptors share a common mode of intracellular signalling: They control the on/off state of a variety of trimeric G proteins (G{alpha}{beta}{gamma}) by stimulating the exchange of GDP for GTP on the {alpha} subunit (G{alpha}). The result is that G{alpha} or G{beta}{gamma}, after dissociation of the trimer, will interact with downstream effectors of the receptor. In the case of the TSH receptor, the main G protein involved is Gs, which activates adenylyl cyclase via Gs{alpha}. In some species, including man, the TSH receptor is also capable of activating phospholipase C (via Gq), thus stimulating the production of diacylglycerol and inositolphosphate (IP{sub 3}). However, higher concentrationsmore » of TSH are required to activate phospholipase C, compared with adenylyl cyclase. As a consequence, the main second messenger of TSH effects on the human thyroid is cyclic AMP. The present review will summarize recent findings identifying mutations of the TSH receptor gene as a cause for thyroid diseases. 59 refs., 4 figs.« less
Article
It has recently been shown that somatic and germ line mutations of the TSH receptor gene cause autonomous hyperfunctioning thyroid adenomas and nonautoimmune toxic thyroid hyperplasia by constitutive activation of the TSH receptor. A "saturated" map of these mutations is a prerequisite for a systematic screening for these clinically important mutations. In this context, it is also of interest to determine whether different amino acid substitutions at the same residue cause constitutive activation of the TSH receptor, as suggested by site-directed mutagenesis of the alpha 1 beta-adrenergic receptor. We, therefore, screened further hyperfunctioning autonomous adenomas of the thyroid for constitutively activating mutations. We identified two new somatic mutations, changing alanine in position 623 to valine (A623V) and threonine in position 632 to isoleucine (T632I). Both mutations constitutively activated cAMP when transiently expressed in COS cells. Together with neighboring mutations, the T632I mutation de...
Article
Unresponsiveness to TSH has been identified and sufficiently studied in only three patients. We report siblings with this defect as the first documentation of familial occurrence. A 26-yr-old woman was diagnosed with congenital hypothyroidism during infancy. The thyroid was atrophic, and thyroid function tests without T4 replacement showed serum free T4 levels below 3 pmol/L, serum TSH of 125 mU/L, and serum thyroglobulin below 5 mg/L. 123I scintigram showed decreased uptake (5% at 24 h), but normal shape at the correct position in the neck. Autoantibodies against thyroglobulin, thyroid peroxidase, and TSH receptor in serum were not detected. The amount of cAMP released into FRTL-5 cell culture in the presence of TSH from the patient was not different from that released by the same amount of TSH from normal subjects, suggesting that TSH bioactivity in our patient was normal. The brother of the patient also had congenital hypothyroidism, and the data on his thyroid function was similar to that for his sist...
Article
Hyperfunctioning thyroid adenomas are clonal neoplasms with the intrinsic capacity of growing and differentiate independently of thyroid-stimulating hormone (TSH). We analysed the mRNA encoding thyrotropin receptor of 11 adenomas obtained by fine needle aspiration biopsy (FNAB) and found 7 mutants all located in three aminoacids clustered in the sixth transmembrane domain of the receptor. These mutations were somatic and specifically present in the tumour tissue. DNA sequence revealed that 80 to 90% of the mutations can be rapidly screened and identified by restriction enzyme analysis of the amplified cDNA obtained from the FNABs. The mutation Thr->Ile was introduced in the wild type receptor and expressed in mouse fibroblasts. These cells constitutively activate the transcription of a reporter gene under the control of cyclic AMP responsive element.
Article
Constitutively activating mutations have recently been identified in the thyrotropin receptor (TSHR) of hyperfunctioning thyroid adenomas and familial hyperthyroidism. In the present study, we evaluated the frequency of constitutively activating TSHR mutations in a large series of autonomously functioning thyroid nodules (AFTNs) in Japan. Forty-five AFTNs (38 solitary hyperfunctioning thyroid adenomas and 7 toxic multinodular goiters) were analyzed. Genomic DNA was extracted from paraffin-embedded tissue sections, from which DNA fragments encoding the mutational hot spots of the receptor (the third cytoplasmic loop and the sixth transmembrane segment) were amplified by polymerase chain reaction. In the single-stranded conformation polymorphism (SSCP) analysis, only one hyperfunctioning adenoma (no. 21) displayed a migration abnormality. In sequence analysis, an unusual mutation of alternate three-base deletions at nucleotides 1953-1957 (AAA GAT ACC to AAG TCC), resulting in one amino acid deletion (Asp at...
Article
THE pituitary hormone thyrotropin stimulates the function, expression of differentiation and growth of thyrocytes by cyclic AMP-dependent mechanisms1-3. Tissue hyperplasia and hyperthyroidism are therefore expected to result when activation of the adenylyl cyclase-cAMP cascade is unregulated. This is observed in several situations4,5, including when somatic mutations impair the GTPase activity of the G protein Gsa (refs 6,7). Such a mechanism is probably responsible for the development of a minority of mono-clonal hyperfunctioning thyroid adenomas6,8,9. Here we identify somatic mutations in the carboxy-terminal portion of the third cytoplasmic loop of the thyrotropin receptor in three out of eleven hyperfunctioning thyroid adenomas. These mutations are restricted to tumour tissue and involve two different residues (aspartic acid at position 619 to glycine in two cases, and alanine at position 623 to isoleucine in one case). The mutant receptors confer constitutive activation of adenylyl cyclase when tested by transfection in COS cells. This shows that G-protein-coupled receptors are susceptible to constitutive activation by spontaneous somatic mutations10,11 and may thus behave as proto-oncogenes.
Article
The gene for human thyrotropin receptor (TSHR) was assigned to chromosome J4, band q31, by in situ hybridization, using a probe for TSHRCopyright © 1990 S. Karger AG, Basel
Article
A human thyroid cDNA library was screened by hybridization with a dog thyrotropin receptor (TSHr) cDNA. Sequencing of the resulting clones identified a 2292 residue open reading frame encoding a 744 amino acid mature polypeptide presenting 90.3% similarity with the dog TSHr. Two major transcripts (4.6 and 4.4 kilobases) were identified in the human thyroid which suggests that alternative splicing could generate multiple forms of human TSHr. Transfection of the coding sequence in COS-7 cells conferred to a membrane preparation of these cells the ability to bind specifically TSH. TSH binding was completely displaced by immunoglobulin preparations from patients with idiopathic myxoedema.